CN108290898B - Quinoline analogs as phosphatidylinositol 3-kinase inhibitors - Google Patents

Quinoline analogs as phosphatidylinositol 3-kinase inhibitors Download PDF

Info

Publication number
CN108290898B
CN108290898B CN201780004233.0A CN201780004233A CN108290898B CN 108290898 B CN108290898 B CN 108290898B CN 201780004233 A CN201780004233 A CN 201780004233A CN 108290898 B CN108290898 B CN 108290898B
Authority
CN
China
Prior art keywords
compound
pi3k
pharmaceutically acceptable
acceptable salt
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Active
Application number
CN201780004233.0A
Other languages
Chinese (zh)
Other versions
CN108290898A (en
Inventor
郝小林
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Nanjing Zhengxiang Pharmaceuticals Co Ltd
Original Assignee
Hangzhou Zheng Xiang Medicine Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Hangzhou Zheng Xiang Medicine Co ltd filed Critical Hangzhou Zheng Xiang Medicine Co ltd
Priority to CN202011068570.6A priority Critical patent/CN112250665A/en
Publication of CN108290898A publication Critical patent/CN108290898A/en
Application granted granted Critical
Publication of CN108290898B publication Critical patent/CN108290898B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Abstract

The present invention provides selective phosphoinositide 3-kinase inhibitors of formula (a) or a pharmaceutically acceptable salt thereof. These compounds are useful for treating conditions mediated by one or more subtypes of P13K (e.g., PI 3K). The invention also provides methods of using these compounds to inhibit phosphoinositide 3-kinase inhibitors to treat conditions associated with phosphatidylinositol 3-kinase activity.

Description

Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
Cross Reference to Related Applications
This application claims the benefit of U.S. provisional patent application serial No. 6/2304148, 2016, 3,4, the contents of which are incorporated herein by reference in their entirety.
Technical Field
The present invention relates generally to quinoline analogs that are inhibitors of phosphatidylinositol 3-kinase (PI3K) activity. More specifically, the invention also relates to the preparation of the disclosed PI3K inhibitor analogs and their use in pharmaceutical compositions for the treatment of various diseases, conditions, and disorders associated with PI3K activity.
Background
Class I phosphoinositide 3-kinases (PI3Ks) regulate phosphatidylinositol 4, 5-bisphosphate (PIP2) phosphorylation. PI3K converts PIP2 into the scaffold binding element phosphatidylinositol (3,4,5) -triphosphate (PIP 3). PIP3 plays a key regulatory role in cell survival, signal transduction, control of membrane trafficking, and other functions. (Di Paolo, G.et al. Nature 2006,443,651; Parker, P.J.et al. biochem. Soc. Trans.2004,32,893; Hawkins, P.T.et al. biochem. Soc. Trans.2006,34,647; Schaeffer, E.M.et al. curr. Opin. Immunol.2000, 12,2822). Dysregulation leads to various disease states such as cancer, inflammation and autoimmune diseases.
Class I PI3Ks is composed of four kinases, which are further divided into 2 subclasses. Class 1A PI3Ks consists of three closely related kinases PI3K α, PI3K β and PI3K, PI3K α, PI3K β and PI3K, which exist as heterodimers, consisting of a catalytic subunit (p110 α, p110 β or p110) and one of several regulatory subunits. Class 1A PI3Ks generally responds to signaling through Receptor Tyrosine Kinases (RTKs). PI3K γ is a single class 1B subtype that responds primarily to G Protein Coupled Receptors (GPCRs) and is composed of the p110 γ catalytic subunit and one of two distinct regulatory subunits. PI3K α and PI3K β are widely expressed in various tissue and organ types. PI3K γ is mainly present in leukocytes, but is also present in skeletal muscle, liver, pancreas and heart (Cantly, c.science 2002,1655). The expression pattern of PI3K is limited by spleen, thymus and peripheral blood leukocytes (Knight, z.et al.cell 2006,125,733).
PI3K has been considered to be a major participant in the adaptive immune system due to its expression pattern and the accumulated evidence of genetically modified mice. Recently, PI3K activation syndrome (APDS) was described as a Primary Immunodeficiency (PID) associated with a dominant gain of function mutation in which lysine was substituted for glutamic acid at residue 1021 in the p110 protein (E1021K). APDS are characterized by repetitive respiratory infections, progressive airway injury, and lymphopenia (Ivan anguloet. science 2013,342,866). PI3K inhibitors could potentially supplement the treatment of B cell related diseases such as Rheumatoid Arthritis (RA) and Systemic Lupus Erythematosus (SLE), as well as Primary Immunodeficiency (PID), with the biological agents rituximab (Rituxan) and belimumab (Benlysta). Several PI3K selective inhibitors, such as idelalisib (GS-1101), IPI-145 and AMG 319, have entered the clinic for hematological malignancies, but few inhibitors have entered clinical trials for anti-inflammatory therapy (Cushing, t.et al.j Med chem.2015,58,480).
7 months 2014, FDA and EMA approved the pioneer PI3K inhibitor idelalisis for the treatment of different types of leukemia; the safety and efficacy of idelalisis treatment for both recurrent FL and recurrent SLL was determined in clinical trials with indolent non-hodgkin lymphoma patients. ("FDA approved Zydeig for three types of hematological cancers", Food and drug Administration, 2014.7.23). However, the us label of idelalisis has a black box warning, indicating that toxicity (including hepatotoxicity) may be severe and fatal. Mortality and/or severe hepatotoxicity occurred in 18% of patients treated with idelalisib monotherapy and in 11% of patients treated with idelalisib in combination trials. The elevation of ALT or AST is greater than 5 times the upper limit of the normal occurrence of ALT or AST. Hepatotoxicity may be associated with inhibition and induction of CYP enzymes by erigeron and its metabolite GS-56363.
(http://www.accessdata.fda.gov/drugsatfda_docs/nda/2014/206545Orig1s000ClinPharmR.pdf)。
Recently, it has been reported that inactivation of mouse p110 prevents a variety of cancers, including non-hematologic solid tumors, and that inactivation of regulatory T cells p110 releases CD8(+) cytotoxic T cells and induces tumor regression. Therefore, p110 inhibitors can break tumor-induced immune tolerance and have potentially broad application in oncology. (Ali, et al, Nature:2014,510, 407-. There remains an unmet need for optimal PI3K inhibitors. For example, the in vivo stability of PI3K inhibitors can be improved, thereby overcoming CYP enzyme inhibition or induction tendencies; an ideal PI3K inhibitor may have the potential to combine malignant tumor treatment with other anticancer interventions, such as emerging immunotherapy. The present invention provides novel compounds, i.e., inhibitors of the PI3K subtype with significantly improved properties.
Disclosure of Invention
Described herein are compounds and pharmaceutically acceptable salts, prodrugs, or solvates thereof, useful for inhibiting the PI3K subtype (e.g., PI 3K). Also provided herein are: compositions, including pharmaceutical compositions containing the compositions; and methods of using and making the compounds. The compounds provided herein are useful for treating diseases, disorders, or conditions mediated by the PI3K subtype, such as PI 3K.
In one aspect, the present invention provides a compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate thereof, wherein:
Figure BDA0001675871200000021
Figure BDA0001675871200000031
x is N or CH;
each R1And R2Independently H, F or SO2Me。
R3Is F or Cl.
In one embodiment of formula (A), where X is N, R1And R2Are all H, R3Is 7-F. The compound is a compound (2) in table 1; or a pharmaceutically acceptable salt, prodrug or solvate thereof.
Figure BDA0001675871200000032
In another embodiment of formula (A), wherein X is C, R1Is H, R2Is 2-SO2Me,R3Is 8-F, which is compound (7) in table 1, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
Figure BDA0001675871200000033
The invention also provides methods of treating a condition or disorder mediated by PI3K with a compound of formula (a), or a pharmaceutically acceptable salt, prodrug, or solvate thereof. The invention further provides methods of treating inflammatory diseases such as asthma, chronic obstructive pulmonary disease, rheumatoid arthritis, multiple sclerosis, and lupus.
The present invention also provides a method of inhibiting the growth or proliferation of a cancer cell, the method comprising contacting the cancer cell with an effective amount of a compound of formula (a), or a pharmaceutically acceptable salt, prodrug, or solvate thereof. The invention also provides a method for increasing the sensitivity of cancer cells to chemotherapy, the method comprising administering to a patient undergoing chemotherapy a chemotherapeutic agent and a compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate, sufficient to increase the sensitivity of cancer cells to the chemotherapeutic agent.
The invention also provides an article of manufacture comprising a compound of formula (a) or a pharmaceutically acceptable salt, or prodrug, or solvate thereof.
Detailed Description
The purpose of which should be understood to be that R1And R2Each and every variation in (a) can be combined with each and every variation in each and every X and X described for formula (a) as if each and every combination were described separately.
PI3K inhibitor compounds
Provided herein are compounds that function as PI3K inhibitors. In one aspect, there is provided a compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate, wherein:
Figure BDA0001675871200000041
x is N or CH;
each R1And R2Independently H, F or SO2Me。
R3Is F or Cl.
In one embodiment of formula (A), where X is N, R1And R2Are all H, R3Is 7-F. The compound is a compound (2) in table 1; or a pharmaceutically acceptable salt, prodrug or solvate thereof.
Figure BDA0001675871200000042
Figure BDA0001675871200000051
In another embodiment of formula (A), wherein X is C, R1Is H, R2Is 2-SO2Me,R3Is 8-F, which is compound (7) in table 1, or a pharmaceutically acceptable salt, prodrug or solvate thereof.
Figure BDA0001675871200000052
TABLE 1 representative quinoline Compounds of formula (A)
Figure BDA0001675871200000053
The present invention also provides a compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate thereof. In certain embodiments, provided herein are crystalline and amorphous forms of a compound of formula (a), or a pharmaceutically acceptable salt, prodrug, or solvate thereof.
"pharmaceutically acceptable salt" refers to salts prepared by conventional methods and well known to those skilled in the art. "pharmacologically acceptable salts" include basic salts of inorganic and organic acids (Berge et al, j.pharm.sci.1977,66: 1).
A "solvate" is formed by treating a compound in a solvent. The present invention also provides solvates of salts of the compounds of formula (a). In the case of treating the compound with water, the solvate is a hydrate. The present invention also provides a hydrate of the compound of formula (a).
"prodrug" includes any compound that is converted to a compound of formula (a) when administered to a subject, for example, after metabolic processing of the prodrug.
Therapeutic use of compounds
A compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate thereof is useful for treating a PI3K mediated disease or condition. In one embodiment, methods of inhibiting PI3K activity using a compound of formula (a) or a pharmaceutically acceptable salt, prodrug, or solvate thereof are provided. In another example, both PI3K and the gamma isomer may be inhibited for optimal efficacy.
In addition to the therapeutic uses described herein, a selected compound of formula (a), or a pharmaceutically acceptable salt, prodrug or solvate thereof, improves the properties of at least one of the following parameters: (i) human hepatocyte stability, and (ii) pharmacokinetic Properties (PK), including oral exposure.
In another embodiment, a selected compound of formula (a), or a pharmaceutically acceptable salt, prodrug, or solvate thereof, has improved human hepatocyte stability. In many cases, the correlation of stability in human hepatocytes to human pharmacokinetics is superior to that of corresponding rodent pharmacokinetic studies. According to some embodiments, the hepatocyte stability of a selected compound is that the half-life may be greater than 24 hours.
As used herein, "treating" or "treatment" with respect to a disorder means alleviating or preventing the disorder or one or more biological manifestations of the disorder to intervene at one or more points in the biological cascade that leads to or is the cause of the disorder, thereby alleviating one or more symptoms or effects associated with the disorder. As noted above, "treatment" of a disorder includes disorder prevention, and "prevention" is understood to refer to prophylactic administration of a drug to significantly reduce the likelihood or severity of a disorder or biological manifestation thereof, or to delay the onset of such a disorder or biological manifestation thereof.
"subject" refers to humans (including adults and children) or other animals. In one embodiment, "patient" refers to a human.
As used herein, a "safe and effective dose" with respect to a compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate thereof is an amount sufficient to treat the symptoms of a patient but low enough to avoid serious side effects. The safe and effective dosage of the compounds will vary with such factors as: the particular compound selected (e.g., taking into account the potency, efficacy, and half-life of the compound); the chosen route of administration; the condition to be treated; the severity of the condition to be treated; the age, volume, weight and physical condition of the patient to be treated; the medical history of the patient to be treated; the duration of the treatment; the nature of concurrent therapy; desired therapeutic effect and the like.
By "inhibition of PI3K activity" or variant is meant a reduction in PI3K activity, relative to the activity of PI3K in the absence of a compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate thereof, and as a direct or indirect response to the presence of a compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate thereof. The term "PI 3K selective inhibitor" generally refers to a compound that more effectively inhibits the activity of the PI3K subtype than other subtypes of the PI3K family (e.g., PI3K α, PI3K β, or PI3K γ).
The efficacy of a compound as an inhibitor of enzyme activity (or other biological activity) can be determined by determining the concentration of each compound that inhibits activity to a predetermined degree, and then comparing the results. The "IC 50" or "IC 90" of an inhibitor can be determined by the concentration that inhibits 50% or 90% of the activity in a biochemical experiment, which can be accomplished using conventional techniques known in the art, including the techniques described in the examples below.
PI3K is expressed primarily in hematopoietic cells including leukocytes such as T cells, dendritic cells, neutrophils, mast cells, B cells, and macrophages. Due to its overall role in immune system function, PI3K is also involved in a number of diseases associated with adverse immune responses, such as allergic reactions, inflammatory diseases, inflammation-mediated angiogenesis, rheumatoid arthritis, autoimmune diseases such as lupus, asthma, emphysema, and other respiratory diseases. By inhibiting the abnormal proliferation of hematopoietic cells, PI3K inhibitors can alleviate symptoms caused by primary effects and secondary symptoms such as systemic or local excess levels of leukocytes or lymphocytes.
In one aspect, the present invention therefore provides a method of treating a condition mediated by inappropriate PI 3-kinase activity, which method comprises administering a safe and effective dose of a compound of formula (a), or a pharmaceutically acceptable salt, prodrug or solvate thereof.
In one embodiment, the PI 3K-mediated disease or condition is selected from: respiratory diseases including asthma, Chronic Obstructive Pulmonary Disease (COPD), and Idiopathic Pulmonary Fibrosis (IPF); allergic diseases (including allergic rhinitis and atopic dermatitis); autoimmune diseases (including rheumatoid arthritis and multiple sclerosis); inflammatory disorders (including inflammatory bowel disease); hematological malignancies; a solid tumor; neurodegenerative diseases; pancreatitis; renal disease; transplant rejection (transplantation rejection); graft rejection (graft rejection); the lungs are injured.
In one embodiment, the compounds described herein may be used to treat cancer mediated by inappropriate PI3K activity. In certain embodiments, the disease is a hematological malignancy. In certain embodiments, the disease is a lymphoma, such as Burkitt's lymphoma (Burkitt lymphoma), diffuse large B-cell lymphoma (DLBCL) and Mantle Cell Lymphoma (MCL), follicular lymphoma, lymphoplasmacytic lymphoma, waldenstrom's macroglobulinemia, and marginal zone lymphoma. In one embodiment, the disorder is multiple myeloma or leukemia, such as Acute Lymphocytic Leukemia (ALL), Acute Myelogenous Leukemia (AML), Chronic Lymphocytic Leukemia (CLL), Small Lymphocytic Lymphoma (SLL), myelodysplastic syndrome (MDS), myeloproliferative disorder (MPD), Chronic Myelogenous Leukemia (CML). In other embodiments, the disease is a solid tumor. In particular embodiments, the indication is treatment of a solid tumor in which PI3K is abnormally expressed, such as pancreatic cancer, gastric cancer, esophageal cancer, and breast cancer. In some embodiments, the compounds, alone or in combination with other anti-cancer therapies, can be used to treat prostate cancer, bladder cancer, colorectal cancer, renal cancer, hepatocellular cancer, lung cancer, ovarian cancer, cervical cancer, head and neck cancer, melanoma, neuroendocrine cancer, brain tumors, bone cancer, or soft tissue sarcoma.
In some embodiments, the PI 3K-mediated disease or disorder is a severe autoimmune disease, such as asthma, type I diabetes, rheumatoid arthritis, multiple sclerosis, Chronic Obstructive Pulmonary Disease (COPD), and lupus.
Combination therapy
In one embodiment, a compound of formula (a), or a pharmaceutically acceptable salt, prodrug, or solvate thereof, can be used in combination with one or more additional therapeutic agents to treat cancer or an inflammatory disorder. The one or more additional therapeutic agents may be a chemotherapeutic agent, radiation therapy, targeted therapy, immunotherapeutic agent, or any of the best care therapies currently available, as small molecules or biological properties. Targeted therapies include, but are not limited to, inhibitors of the following kinases: cyclin Dependent Kinases (CDKs) such as CDK1, CDK2, CDK4/6, CDK7 and CDK 9; janus kinases (JAKs) such as JAK1, JAK2, and/or JAK3, spleen tyrosine kinase (SYK); bruton's Tyrosine Kinase (BTK); mitogen-activated protein kinases such as MEK1 and MEK 2; bromodomain-containing protein inhibitors (BRDs) such as BRD 4; isocitrate Dehydrogenases (IDHs) such as IDH1, Histone Deacetylases (HDACs), or any combination thereof.
Chemotherapeutic agents can be classified by their mechanism of action as: alkylating agents, antimetabolites, antimicrotubule agents, topoisomerase inhibitors, and cytotoxic agents. A compound of formula (a), or a pharmaceutically acceptable salt, prodrug or solvate thereof, can be used in combination with chemotherapy to sensitize and improve the efficacy of certain chemotherapeutic agents for the treatment of blood or solid tumors.
Immunotherapeutics include, but are not limited to, therapeutic antibodies, small molecules and vaccines suitable for treating patients; such as IDO1 and TDO2 inhibitors, A2A receptor inhibitors, arginase inhibitors, toll-like receptor agonists, chemokine modulators (including the CCR and CXCR family), checkpoint blocking antibodies such as antibodies that modulate PD-1, PD-L1, CTLA-4, OX40-OX40 ligand, LAG3, TIM3, or any combination thereof.
Radiation therapy is part of cancer treatments that control or kill malignant cells, and is commonly applied to cancerous tumors due to its ability to control cell growth. The compound of formula (a) or a pharmaceutically acceptable salt, prodrug or solvate thereof may be used in combination with radiation therapy to improve the efficacy of radiation therapy for treating blood or solid tumors, or in combination with surgery, chemotherapy, immunotherapy and four methods.
In certain embodiments, a compound of formula (a), or a pharmaceutically acceptable salt, prodrug, or solvate thereof, may be used in combination with one or more additional therapeutic agents to treat patients who are substantially refractory to treatment with at least one chemotherapeutic treatment or who relapse after treatment with a chemotherapeutic treatment.
Pharmaceutical composition
In another aspect, the present invention provides a pharmaceutical composition comprising a compound of formula (a), or a pharmaceutically acceptable salt, prodrug, or solvate salt thereof, and a pharmaceutically acceptable carrier or excipient. The pharmaceutical compositions may be formulated for specific routes of administration, such as oral, parenteral, topical, and the like.
Compositions intended for oral use are prepared according to any method known in the art for the preparation of pharmaceutical compositions, and can be prepared in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups, and capsules. Oral compositions may contain the active ingredient in admixture with pharmaceutically acceptable non-toxic excipients which are suitable for the manufacture of tablets. The tablets are uncoated or coated by known techniques to delay disintegration and absorption in the gastrointestinal tract and thereby provide a sustained action over a longer period. Formulations for oral use may be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert solid diluent, for example calcium carbonate, calcium phosphate or kaolin, or as soft gelatin capsules wherein the active ingredient is mixed with water or an oil medium, for example peanut oil, liquid paraffin or olive oil.
Certain injectable compositions are aqueous isotonic solutions or suspensions, and suppositories are advantageously prepared from fatty emulsions or suspensions. Suitable compositions for transdermal administration include an effective amount of a compound of the present invention with a suitable carrier. Suitable carriers for transdermal delivery include absorbable pharmacologically acceptable solvents to aid passage through the skin of the host. For example, the transdermal device is in the form of a bandage comprising: a backing member; a container containing the compound and optionally a carrier; an optional rate control barrier for delivering the compound to the skin of the host at a controlled and predetermined rate over an extended period of time; and means for securing the device to the skin.
Suitable compositions for topical application (e.g., to the skin and eye) include aqueous solutions, suspensions, ointments, creams, gels, or spray formulations, for example, by delivery via aerosol or the like. Such topical delivery systems are particularly suitable for dermal application, e.g. for the treatment of skin cancer, e.g. for prophylactic use in sunscreens, lotions, sprays and the like.
Topical administration as used herein may also involve inhalation or intranasal administration. The compositions of the present invention may conveniently be delivered in dry powder form (either as a mixture alone, for example a dry blend with lactose, or as particles of a mixed component, for example with a phospholipid), from a dry powder inhaler, or in the form of an aerosol spray presentation from a pressurised container, pump, spray, atomising nozzle or atomiser (with or without the use of a suitable propellant).
The invention further provides pharmaceutical compositions and dosage forms comprising one or more agents that reduce the rate of decomposition of a compound of the invention as an active ingredient. Such agents, referred to herein as "stabilizers," include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers, and the like.
Modes of administration and dosages
The pharmaceutical compositions may be administered in single or multiple doses. The compound of formula (a), or a pharmaceutically acceptable salt, prodrug or solvate salt thereof, may be formulated to provide a desired release profile of the active ingredient for therapeutic treatment purposes.
The pharmaceutical compositions are preferably prepared in the form of dosage units containing the active ingredient in a specific amount in the form of tablets, pills, powders, suspensions, emulsions, solutions, syrups and capsules. For example, these pharmaceutical compositions may contain the active ingredient in an amount of about 0.1mg to 1000mg, preferably about 0.1mg to 500 mg. Suitable daily dosages for humans or other mammals can vary widely depending on the patient's symptoms and other factors, but can again be determined using routine methods. The daily dose may be 1 to 4 doses administered per day. For therapeutic purposes, the active compounds of the invention are usually combined with one or more adjuvants (drops suitable for the intended route of administration), which are suitable for administration to the eye, ear or nose. Suitable topical dosages of the active ingredient of the compounds of the invention are from 0.1mg to 150mg, administered from 1 to 4 times per day, preferably 1 or 2 times. For topical administration, the active ingredient may be present at 0.001% w/w to 10% w/w, for example 1% w/w to 2% w/w, preferably not more than 5% w/w, more preferably 0.1% to 1% by weight of the formulation.
In particular embodiments, the method comprises administering to the subject an initial daily dose of about 0.1mg to 500mg of the compound of formula (a), and increasing the dose by increments until clinical efficacy is achieved. The dosage may be increased using increments of about 5mg, 10mg, 25mg, 50mg or 100 mg. The amount may be increased daily, every other day, twice weekly or once weekly. Synthesis of Compound of formula (A)
The compounds of formula (a) may be prepared using the methods disclosed herein and conventional variations thereof, as will be apparent in light of the disclosure herein, and the methods of preparation are well known in the art. Conventional and well-known synthetic methods may be used in addition to the teachings herein. The synthesis of representative compounds described herein can be accomplished as described in the examples below. Reagents may be purchased commercially, for example from Sigma Aldrich or other chemical suppliers, if available.
Overview
The reagents and solvents used below are available from commercial sources. Recording on Mercury 300MHZNMR spectrometer
1H-NMR spectrum. The important peaks are listed in the following order: multiplicity (s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; br s, broad singlet), the coupling constants being expressed in Hertz (HZ) and the number of protons. Mass spectrometry results are reported as the ratio of mass to charge, followed by the relative abundance of each ion (in parentheses). Electron Spray Ionization (ESI) mass spectrometry was performed on an Agilent 1100 series LC/MSD electrospray mass spectrometer. All compounds were able to be analyzed in positive ESI mode using acetonitrile in water with 0.1% TFA as the delivery solvent.
Synthesis reaction
The terms "solvent", "inert organic solvent" or "inert solvent" refer to a solvent that is inert under the reaction conditions described in connection therewith (including, for example, benzene, toluene, acetonitrile, tetrahydrofuran ("THF"), dimethylformamide ("DMF"), chloroform, Dichloromethane (DCM), diethyl ether, methanol, pyridine, and the like). Unless stated to the contrary, the solvent used for the reaction of the present invention is an inert organic solvent, and the reaction is carried out under an inert gas, preferably nitrogen.
Preparation of 8-substituted quinolinamines:
Figure BDA0001675871200000111
example 1: (S) -1- (8-fluoro-2-pyridin-2-yl-quinolin-3-yl) -ethylamine (9)
Step 1:
Figure BDA0001675871200000112
to a solution of 2-chloro-8-fluoroquinoline-3-carbaldehyde (1.5g, 7.2mmol) in anhydrous THF (20mL) at room temperature was added titanium isopropoxide (4.3mL, 1.4 mmol). After 15 min, (R) -2-methyl-2-propanesulfinamide (0.867g, 7.2mmol) was added and stirring continued at room temperature overnight. Water (100mL) was added to the reaction mixture, and the resulting precipitate was filtered and washed with DCM. The organic layer was dried (Na)2SO4) Filtered and concentrated in vacuo to give the starting material as a pale yellow solid which was purified by silica gel column chromatography (ethyl acetate/hexanes, 4/5) to give a pale yellow solid (2.0 g, 89%). Mass Spectrometry (ESI) m/e: 313(M +1).
Step 2:
Figure BDA0001675871200000121
to a solution of 2-methyl-propane-2-sulfinic acid 2-chloro-8-fluoro-quinolin-3-ylmethyleneamide (0.95g, 2.8mmol) in DCM (22mL) was added MeMgCl (1.94mL, 5.8 mmol; 3M in THF solution) dropwise at-78 deg.C under nitrogen for 10 minutes. The reaction mixture was allowed to reach room temperature and stirred overnight. Slowly add saturated NH under stirring4Aqueous Cl (50mL), mixture cooled in ice salt, aqueous layer extracted with DCM (2 × 50mL), organic layer dried (MgSO 50)4) Filtered and concentrated in vacuo to give a yellow oil which was purified by silica gel column chromatography (ethyl acetate/hexane, from 4/5 to 100% ethyl acetate) to give a pale yellow solid (260mg, 28%). Mass Spectrometry (ESI) M/e:329(M +1).
And step 3:
Figure BDA0001675871200000122
in N2(S) -2-methyl-propane-2-sulfinic acid [1- (2-chloro-8-fluoro-quinolin-3-yl) -ethyl ] -in dioxane (5mL)]Amide (0.186g, 0.57mmol), Pd (PPh)3)4A mixture of (0.066g, 0.057mmol, 0.1 equiv.) and 2- (tributylstannyl) -pyridine (0.51g, 1.4mmol, 2.4 equiv.) was heated to 110 ℃. After stirring overnight, the combined solvents were concentrated and purified by silica gel column chromatography (ethyl acetate) to give 2-methyl-propane-2-sulfinic acid [1- (8-fluoro-2-pyridin-2-yl-quinolin-3-yl) -ethyl]Amide (160mg, 43%). Mass Spectrometry (ESI) M/e:372(M +1).
To a solution of the above material (160mg) in MeOH (2mL) was added 4N HCl in dioxane (2mL) at room temperature, and the resulting reaction mixture was stirred for 2 hours and concentrated under reduced pressure. Diethyl ether was added and sonicated for 2 minutes, filtered to give (S) -1- (8-fluoro-2-pyridin-2-yl-quinolin-3-yl) -ethylamine as the HCl salt. Mass Spectrometry (ESI) M/e:268(M +1).
In a similar manner (S) -1- (8-chloro-2-pyridin-2-yl-quinolin-3-yl) -ethylamine (10) and (S) -1- [ 8-fluoro-2- (2-methanesulfonyl-phenyl) -quinolin-3-yl ] -ethylamine (11) were prepared.
Preparation of 7-substituted quinolinamines:
Figure BDA0001675871200000131
example 2: (S) -1- (7-fluoro-2-phenyl-quinolin-3-yl) -ethylamine (12)
Figure BDA0001675871200000132
The compound (S) -2- (1- (2-chloro-7-fluoroquinolin-3-yl) ethyl) isoindoline-1, 3-dione was prepared according to the literature (j.med.chem.2015,58, 480-511). In N2This compound (280mg, 0.79mmol), phenylboronic acid (146mg, 1.2mmol) and potassium carbonate (328mg, 2.4mmol) were combined under atmosphere in 6mL anhydrous DMF. After addition of PdCl2(dppf) before DCM (64mg, 0.079mmol), the solution was treated with N2Purge about 5 minutes. The solution was heated at 100 ℃ for 3 hours and then cooled to 50 ℃. Will dissolveThe solution was concentrated in vacuo to give a brown residue, which was diluted with ethyl acetate (12mL), the organic layer was washed with water (3 × 3mL), followed by brine (10mL), the combined aqueous layers were extracted with DCM (3 × 2mL), MgSO 24The combined organic layers were dried and then concentrated in vacuo. The resulting residue was purified by flash chromatography on silica gel eluting with a gradient of 20% to 40% ethyl acetate in hexanes. The fractions containing the pure product were combined and concentrated in vacuo to give (S) -2- [1- (7-fluoro-2-phenyl-quinolin-3-yl) -ethyl]Isoindole-1, 3-dione (263mg, 84% yield) as a pale yellow foam. Mass Spectrometry (ESI) M/e:397(M +1).
To (S) -2- [1- (7-fluoro-2-phenyl-quinolin-3-yl) -ethyl]-isoindole-1, 3-dione (260mg, 0.65mmol) in anhydrous ethanol (3mL) as a slurry and adding NH dropwise2NH2(0.11g, 5.0 equiv.). The reaction mixture was heated to 90 ℃, held for 30 minutes, and cooled to room temperature. The reaction mixture was filtered and washed with ethyl acetate. The resulting ethyl acetate solution was washed with water, brine and Na2SO4And (5) drying. Removal of the solvent gave (S) -1- (7-fluoro-2-phenyl-quinolin-3-yl) -ethylamine as a tan oil (122mg, 71%). Mass Spectrometry (ESI) M/e:267(M +1).
In a similar manner, (S) -1- [2- (3, 5-difluoro-phenyl) -7-fluoro-quinolin-3-yl ] -ethylamine (14), (S) -1- [ 7-fluoro-2- (2-methanesulfonyl-phenyl) -quinolin-3-yl ] -ethylamine (15), and (S) -1- [ 7-fluoro-2- (3-fluoro-phenyl) -quinolin-3-yl ] -ethylamine (16) were prepared.
Example 3: (S) -1- (7-fluoro-2- (pyridin-2-yl) quinolin-3-yl) ethylamine (13)
Figure BDA0001675871200000141
In N2(S) -2- (1- (2-chloro-7-fluoroquinolin-3-yl) ethyl) isoindoline-1, 3-dione (0.86g, 2.4mmol), Pd (PPh) in dioxane (30mL)3)4A mixture of (0.28g, 0.24mmol, 0.1 equiv.) and 2- (tributylstannyl) -pyridine (1.07g, 2.9mmol, 1.2 equiv.) was heated to 90 ℃. After stirring overnight, LC-MS showed 30% completion. The reaction mixture was heated to 101 ℃ and held for an additional 2 days.The reaction mixture was then cooled to room temperature and the resulting solid was filtered and washed with ethyl acetate to give 2- ((S) -1- (7-fluoro-2- (pyridin-2-yl) quinolin-3-yl) ethyl) isoindoline-1, 3-dione (0.84g, 88%) as a tan solid. Mass Spectrometry (ESI) M/e:398(M +1).
To a slurry of 2- ((S) -1- (7-fluoro-2- (pyridin-2-yl) quinolin-3-yl) ethyl) isoindoline-1, 3-dione (0.84g, 2.1mmol) in anhydrous ethanol (5mL) was added dropwise NH2NH2(0.34 g, 10.4 mmol). The reaction mixture was heated to 90 ℃, held for 30 minutes, and cooled to room temperature. The reaction mixture was filtered and washed with ethyl acetate. The resulting ethyl acetate solution was washed with water, brine and Na2SO4And (5) drying. Removal of the solvent gave (S) -1- (7-fluoro-2- (pyridin-2-yl) quinolin-3-yl) ethylamine as a tan oil (399mg, 71%). Mass Spectrometry (ESI) m/e:268(M +1).
Preparation of 2,4, 6-triamino-pyrimidine-5-carbonitrile
Figure BDA0001675871200000142
Ammonium hydroxide (4mL) was added to a solution of 2,4, 6-trichloropyrimidine-5-carbonitrile (1.0g, 4.8mmol) in dioxane (4mL) at room temperature. The solution was warmed to 50 ℃ and stirred for 3 hours. The reaction mixture was cooled to 10 ℃ and water (10mL) was added. The resulting solid was filtered, washed with water, and dried under high vacuum to give 2,4, 6-triamino-pyrimidine-5-carbonitrile as a white solid (0.8 g). Mass Spectrometry (ESI) M/e:170(M + H).
(S) -2, 4-diamino-6- {1- [ 7-fluoro-2- (3-fluoro-phenyl) -quinolin-3-yl ] -ethylamino } -pyrimidine-5-carbonitrile (5)
Figure BDA0001675871200000151
Potassium fluoride (34mg, 0.58mmol) was added to a solution of (S) -1- [ 7-fluoro-2- (3-fluoro-phenyl) -quinolin-3-yl ] -ethylamine (90mg, 0.31mmol) and 2, 4-diamino-6-chloropyrimidine-5-carbonitrile (60mg, 0.35mmol) in diisopropylethylamine (0.1mL, 0.60mmol) and DMSO (2 mL). The resulting mixture was heated to 100 ℃ for 14 hours, then the reaction was cooled to room temperature, diluted with water (5mL), the resulting solid was filtered, washed with water, dried and purified by preparative TLC to give (S) -2, 4-diamino-6- {1- [ 7-fluoro-2- (3-fluoro-phenyl) -quinolin-3-yl ] -ethylamino } -pyrimidine-5-carbonitrile as a white solid. Mass spectrum (ESI) M/e 418(M +1).1H NMR (300MHz, DMSO-d6) ppm 8.49(s,1H),8.10(dd, J ═ 5.7,3.0Hz,1H),7.75(dd, J ═ 11,2.4Hz,1H),7.52-7.60(M,3H),7.27-7.32(M,1H),7.12(d, J ═ 7.2Hz,1H),6.52(s,2H),6.10(s, br,2H),5.40-5.45(M,1H),1.30(d, J ═ 6.9Hz, 3H).
The following compounds were prepared in a similar manner.
(S) -2, 4-diamino-6- [1- (7-fluoro-2-phenyl-quinolin-3-yl) -ethylamino ] -pyrimidine-5-carbonitrile (Compound 1, Table 1). Mass Spectrometry (ESI) M/e:400(M +1).1H NMR (300MHz, DMSO-d6) ppm 8.46(s,1H),8.07(dd, J ═ 6.6,2.4Hz,1H),7.67-7.70(m,2H),7.49-7.52(m,3H),7.13(d, J ═ 7.5Hz,1H),6.52(s,2H),6.10(s, br,2H),5.45-5.50(m,1H),1.27(d, J ═ 6.9, 3H).
(S) -2, 4-diamino-6- [1- (7-fluoro-2-pyridin-2-yl-quinolin-3-yl) -ethylamino ] -pyrimidine-5-carbonitrile (Compound 2, Table 1). Mass Spectrometry (ESI) M/e:401(M +1).1H NMR (300MHz, DMSO-d6) ppm 8.72(s,1H),8.56(m,1H),7.54-8.12(m,6H),6.50(s,2H),6.08(s, br,2H),5.65-5.75(m,1H),1.35(d, J ═ 6.9Hz, 3H).
(S) -2, 4-diamino-6- {1- [2- (3, 5-difluoro-phenyl) -7-fluoro-quinolin-3-yl ] -ethylamino } -pyrimidine-5-carbonitrile (Compound 3, Table 1). Mass spectrum (ESI) M/e 436(M +1).1H NMR (300MHz, DMSO-d6) ppm 8.52(s,1H),8.13(dd, J ═ 5.7,3.0Hz,1H),7.76(dd, J ═ 7.5,1.8Hz,1H),7.54-7.60(M,1H),7.08-7.45(M,2H),7.09(d, J ═ 6.9Hz,1H),6.52(s,2H),6.12(s, br,2H),5.35-5.40(M,1H),1.34(d, J ═ 6.3Hz, 3H).
(S) -2, 4-diamino-6- {1- [ 7-fluoro-2- (2-methanesulfonyl-phenyl) -quinolin-3-yl ] -ethylamino } -pyrimidine-5-carbonitrile (Compound 4, Table 1). Mass Spectrometry (ESI) M/e:478(M +1).1H NMR (300MHz, DMSO-d6) ppm 8.50(s,1H),8.13(dd, J ═ 5.7,3.0Hz,1H),7.77-7.89(m,4H),7.56-7.61(m,1H),7.06(d, J ═ 7.5Hz,1H),6.54(s,2H),6.24(s, br,2H),5.15-5.25(m,1H),3.37(s,3H),1.27(d, J ═ 6.9Hz, 3H).
(S) -2, 4-diamino-6- [1- (8-fluoro-2-pyridin-2-yl-quinolin-3-yl) -ethylamino ] -pyrimidine-5-carbonitrile (Compound 6, Table 1). Mass Spectrometry (ESI) M/e:401(M +1).1H NMR (300MHz, DMSO-d6) ppm 8.51(s,1H),8.16(m,1H),7.54-8.02(m,6H),6.51(s,2H),6.01(s, br,2H),5.23-5.34(m,1H),1.30(d, J ═ 6.9Hz, 3H).
(S) -2, 4-diamino-6- {1- [ 8-fluoro-2- (2-methanesulfonyl-phenyl) -quinolin-3-yl ] -ethylamino } -pyrimidine-5-carbonitrile (Compound 7, Table 1). Mass Spectrometry (ESI) M/e:478(M +1).1H NMR (300MHz, MeOH-d4) ppm 8.74(d, J ═ 4.5Hz,1H),8.51(s,1H),8.03-7.94(m,2H),7.80(d, J ═ 8.4Hz,1H),7.61-7.45(m,4H),5.84(q, J ═ 6.9Hz,1H),4.60(s,1H),3.33(s,3H),1.41(d, J ═ 6.9Hz, 3H).
(S) -2, 4-diamino-6- [1- (8-chloro-2-pyridin-2-yl-quinolin-3-yl) -ethylamino ] -pyrimidine-5-carbonitrile (Compound 8, Table 1). Mass Spectrometry (ESI) M/e:417(M +1).1H NMR (300MHz, DMSO-d6) ppm 8.74(d, J ═ 4.5Hz,1H),8.60(s,1H),8.06-7.94(m,3H),7.75(d, J ═ 8.4Hz,1H),7.64-7.53(m,2H),6.48(s,2H),5.80-5.74(m,1H),1.39(d, J ═ 6.0Hz, 3H).
Biological examples
Activity tests were performed in the following examples using the methods described herein and methods well known in the art.
Characterization of the Compound of formula (A)
This example compares the biological activity and hepatocyte stability of a compound of formula (a) with, for example, the 4-amino-2-hydropyrimidine analogue D-F (compound D-F is reported in patent US2013/0267524, compound D being an approximate analogue for reference purposes) having the following structure.
Figure BDA0001675871200000171
The enzymatic activity of different PI3K subtypes was measured to compare the selectivity of the tested compounds for the PI3K subtype, in particular for PI 3K. Hepatocyte stability was also measured to assess the potential half-life of the tested compounds in human subjects.
Each of these biological experiments is described below.
Enzymatic activity of PI3K subtype
The enzymatic activity of class I PI3K subtype in the presence of the compound of table 1 above was measured using a time-resolved fluorescence resonance energy transfer (TR-FRET) assay.
The TR-FRET assay can monitor the formation of the product inositol 3,4, 5-triphosphate molecule (PIP3) because the product inositol 3,4, 5-triphosphate molecule competes with fluorescently labeled PIP3 for binding to the GRP-1 pleckstrin homeodomain protein. When the labeled fluorophore is displaced from the GRP-1 protein binding site, the increase in phosphatidylinositol 3-phosphate product results in a decrease in TR-FRET signal.
PI3K subtype was analyzed at initial rate conditions in the presence of 10 μ MATP and compounds were tested in 10-dose IC50 pattern starting at 0.5 μ M concentration. The control compound PI-103 was tested with 10 doses of IC50, with 3-fold serial dilutions starting at 10 μ M.
Data were normalized on the basis of negative (DMSO) controls. IC50 values for α, β, and γ were calculated from a four parameter equation fitted to the dose-response curve. IC50 is reported in nM.
IC50 values were obtained for all PI3K subtypes (α, β, and γ), and table 2 summarizes the IC50 data for PI3K collected in this example.
Hepatocyte stability
Metabolic stability of Test Article (TA) after incubation in cryopreserved hepatocytes was assessed using a hepatocyte assay to monitor maternal drug disappearance by LC/MC. TA at a final DMSO concentration of 1% was incubated in duplicate with 0.5 million hepatocytes/ml (1. mu.M substrate). At 37 ℃ 5% CO2And incubation under saturated humidity conditions. Samples were taken at 0,1, 2 and 3 hours to monitor the disappearance of TA and determine half-life (t 1/2). Table 2 below summarizes the t1/2 values (e.g., t1/2) collected from this example.
Table 2.
Compound (I) IC50(nM) Hepatocyte T1/2(h)
1 0.043 27
2 0.086 53
3 0.24 17
4 0.27 6
5 0.14 20
6 - 9
7 2.1 59
8 0.97 -
D - 10
E - 4.8
H - 0.88
G - 1.2
F - 5.9
The results of this example demonstrate that compounds 1 and 4 of formula (a) have moderately improved stability (i.e., longer half-life) in human hepatocytes compared to compounds D and E (2.7x and 1.3 x). However, in human hepatocytes, several compounds show a surprisingly significant improvement in stability (i.e., longer half-life); table 3 below shows a comparison of t1/2 for compounds 2, 5,7, H, G and F. In human hepatocytes, significant improvements in compound stability may reduce the formation of drug metabolites in vivo and may positively impact compound safety in clinical trials.
TABLE 3
Figure BDA0001675871200000191
Figure BDA0001675871200000192
Rats were administered compound 2 both intravenously (iv) and orally (PO). Table 4 shows a comparison of PK profiles for compound 2 and a commercial PI3K inhibitor idelalisis (also known as CAL-101(PK data from idelalisis NDA document)): for SD rats, compound 2 showed a significant increase in oral exposure after administration of an oral dose (10 mg/kg). The oral exposure corrected with dose (AUCinf/dose) was 20-fold higher than Idelalisib.
TABLE 4
Figure BDA0001675871200000193
Figure BDA0001675871200000201
TABLE 1 cytotoxicity assay of Compound 2 on selected cell lines
Process for producing a metal oxide
Day 0: cell seeding was performed with all cell lines at a density of 10000 cells/80 μ L/well.
Day 1, drug treatment: a 3-fold serial dilution of the compound stock solution was prepared with growth medium. 20 μ L (5 ×) of drug solution (each concentration in duplicate) was dispensed in each well, with a final DMSO concentration of 0.1% for each well treated with test compound and corresponding vehicle control. The plates were incubated in the indicated incubator for 72 hours.
Day 4, plate reading: the CTG solution was thawed and equilibrated to room temperature, 100 μ LCTG added to each well, and the contents were mixed on a plate shaker for 2 minutes and incubated for 10 minutes before recording the luminescence signal using Envision.
Data analysis
Using a sigmoidal dose-response nonlinear regression model, cell viability for each dose was fitted to GraphPad Prism version 5 (control percentage T/C, optical density of test wells after exposure to test drug for a period of 3 days T, control optical density C) and the IC50 of the test drug concentration was calculated at 100 × T/C50.
TABLE 5 IC50(μM)
Figure BDA0001675871200000202

Claims (6)

1. A selective phosphoinositide 3-kinase (PI3K) inhibitor compound, or a pharmaceutically acceptable salt thereof, the phosphoinositide 3-kinase (PI3K) inhibitor compound having the structure of formula (a):
Figure FDA0002545567560000011
wherein X is selected from N or CH;
R1and R2Each of which is independently selected from H, F and SO2Me, and
R3is 7-F.
2. The selective phosphoinositide 3-kinase (PI3K) inhibitor compound or pharmaceutically acceptable salt thereof according to claim 1, wherein X is N, R1And R2Each of which is H.
3. The selective phosphoinositide 3-kinase (PI3K) inhibitor compound or pharmaceutically acceptable salt thereof according to claim 1, wherein X is CH and R is1Is SO2Me。
4. The selective phosphoinositide 3-kinase (PI3K) inhibitor compound or pharmaceutically acceptable salt thereof according to claim 1, wherein X is CH and R is1Is F.
5. The selective phosphoinositide 3-kinase (PI3K) inhibitor compound or pharmaceutically acceptable salt thereof according to claim 1, wherein X is CH and R is1Is H and R2Is H.
6. Use of a selective phosphoinositide 3-kinase (PI3K) inhibitor compound according to any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for selectively inhibiting the growth or proliferation of phosphoinositide 3-kinase (PI 3K).
CN201780004233.0A 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors Active CN108290898B (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202011068570.6A CN112250665A (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201662304148P 2016-03-05 2016-03-05
US62/304,148 2016-03-05
PCT/US2017/019970 WO2017155741A1 (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CN202011068570.6A Division CN112250665A (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Publications (2)

Publication Number Publication Date
CN108290898A CN108290898A (en) 2018-07-17
CN108290898B true CN108290898B (en) 2020-09-08

Family

ID=59789651

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202011068570.6A Pending CN112250665A (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
CN201780004233.0A Active CN108290898B (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Family Applications Before (1)

Application Number Title Priority Date Filing Date
CN202011068570.6A Pending CN112250665A (en) 2016-03-05 2017-02-28 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Country Status (8)

Country Link
US (2) US10792283B2 (en)
EP (1) EP3400227B1 (en)
JP (1) JP7201992B2 (en)
KR (2) KR20210059033A (en)
CN (2) CN112250665A (en)
CA (2) CA3124815A1 (en)
ES (1) ES2865424T3 (en)
WO (1) WO2017155741A1 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20210059033A (en) 2016-03-05 2021-05-24 항저우 정샹 파마슈티컬즈 컴퍼니 리미티드 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
TW202302566A (en) * 2021-03-09 2023-01-16 大陸商南京征祥醫藥有限公司 Salt or crystalline form of quinoline compound and method for preparing the same and use thereof
CN114276329B (en) * 2021-03-09 2022-11-11 南京征祥医药有限公司 Crystal form of salt of quinoline compound, preparation method and application thereof
CN114276326B (en) * 2021-03-09 2022-11-11 南京征祥医药有限公司 Salt of quinoline compound, preparation method and application

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008118455A1 (en) * 2007-03-23 2008-10-02 Amgen Inc. δ3- SUBSTITUTED QUINOLINE OR QUINOXALINE DERIVATIVES AND THEIR USE AS PHOSPHATIDYLINOSITOL 3-KINASE ( PI3K) INHIBITORS
CN104395305A (en) * 2012-04-04 2015-03-04 安姆根有限公司 Heterocyclic compounds and their uses
US20160016915A1 (en) * 2012-12-21 2016-01-21 Gilead Calistoga Llc Phosphatidylinositol 3-kinase inhibitors

Family Cites Families (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT2137186E (en) * 2007-03-23 2016-03-30 Amgen Inc Heterocyclic compounds and their uses
US20110135655A1 (en) 2009-01-13 2011-06-09 PHILADELPHIA HEALTH AND EDUCATION CORPORATION d/b/a Drexel University College of Medicine; Role of PI3K p110 delta Signaling in Retroviral Infection and Replication
MX2011013510A (en) * 2009-06-25 2012-03-07 Amgen Inc Polycyclic derivatives of pyridine and their use in the treatment of (inter alia) rheumatoid arthritis and similar diseases.
AU2011302196B2 (en) 2010-09-14 2016-04-28 Exelixis, Inc. Inhibitors of PI3K-delta and methods of their use and manufacture
MX2013005005A (en) * 2010-11-04 2013-10-25 Amgen Inc 5 -cyano-4, 6 -diaminopyrimidine or 6 -aminopurine derivatives as pi3k- delta inhibitors.
UY34893A (en) * 2012-07-10 2014-02-28 Takeda Pharmaceutical DERIVATIVES OF AZAINDOL
TWI644909B (en) 2013-06-14 2018-12-21 基利科學股份有限公司 Phosphatidylinositol 3-kinase inhibitors
EP3074016A2 (en) 2013-11-26 2016-10-05 Gilead Sciences, Inc. Therapies for treating myeloproliferative disorders
WO2015106014A1 (en) * 2014-01-09 2015-07-16 Takeda Pharmaceutical Company Limited Azaindole derivatives
US9637488B2 (en) * 2015-01-29 2017-05-02 Fuqiang Ruan Heterocyclic compounds as inhibitors of class I PI3KS
KR20210059033A (en) 2016-03-05 2021-05-24 항저우 정샹 파마슈티컬즈 컴퍼니 리미티드 Quinoline analogs as phosphatidylinositol 3-kinase inhibitors

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008118455A1 (en) * 2007-03-23 2008-10-02 Amgen Inc. δ3- SUBSTITUTED QUINOLINE OR QUINOXALINE DERIVATIVES AND THEIR USE AS PHOSPHATIDYLINOSITOL 3-KINASE ( PI3K) INHIBITORS
CN104395305A (en) * 2012-04-04 2015-03-04 安姆根有限公司 Heterocyclic compounds and their uses
US20160016915A1 (en) * 2012-12-21 2016-01-21 Gilead Calistoga Llc Phosphatidylinositol 3-kinase inhibitors

Also Published As

Publication number Publication date
CN108290898A (en) 2018-07-17
CA3013490A1 (en) 2017-09-14
CN112250665A (en) 2021-01-22
EP3400227B1 (en) 2021-03-31
JP2019511452A (en) 2019-04-25
KR102256497B1 (en) 2021-05-26
CA3124815A1 (en) 2017-09-14
KR20180104160A (en) 2018-09-19
US11446301B2 (en) 2022-09-20
US20210046075A1 (en) 2021-02-18
CA3013490C (en) 2021-09-14
US10792283B2 (en) 2020-10-06
WO2017155741A1 (en) 2017-09-14
US20190365752A1 (en) 2019-12-05
KR20210059033A (en) 2021-05-24
JP7201992B2 (en) 2023-01-11
EP3400227A4 (en) 2019-06-19
ES2865424T3 (en) 2021-10-15
EP3400227A1 (en) 2018-11-14

Similar Documents

Publication Publication Date Title
KR102527784B1 (en) Heterocyclic amides useful as protein modulators
JP6869947B2 (en) Use of substituted quinazoline compound and its G12C mutant KRAS, HRAS and / or NRAS protein as an inhibitor
US11446301B2 (en) Quinoline analogs as phosphatidylinositol 3-kinase inhibitors
EP3960735B1 (en) Quinazoline derivatives used to treat hiv
CN109369671B (en) Fused tricyclic ureas as Raf kinase and/or Raf kinase dimer inhibitors
EP2699573B1 (en) 7-(3,5-dimethyl-4-isoxazolyl)-8-(methyloxy)-1h-imidazo[4,5-c]quinoline derivatives
TW201927771A (en) Heterocyclic amides useful as protein modulators and methods of using the same
BR112020006780A2 (en) modulators of the interferon gene stimulator (sting)
US11542262B2 (en) Phosphatidylinositol 3-kinase inhibitors
US10611777B2 (en) Imidazopyridazine compounds and their use
JP2013542966A (en) Pyrazolopyridines and their use as TYK2 inhibitors and their use
TW201217387A (en) Azabenzothiazole compounds, compositions and methods of use
JP2022502427A (en) Treatment of HPK1-dependent disorders such as cinnoline compounds and cancer
US10793575B2 (en) Oxoisoquinoline derivatives
WO2021004421A1 (en) Alkyne derivative, preparation method for same, and uses thereof
KR20140082710A (en) Imidazopyridine compounds, compositions and methods of use
JP2021506809A (en) 6- (Morpholine-4-yl) -pyridin-2-yl-1H-pyrrolo [3,2-B] pyridine derivative as an mTOR inhibitor

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
TA01 Transfer of patent application right

Effective date of registration: 20180809

Address after: 310000 2 A0101 room 452, 6 Avenue, Baiyang street, Hangzhou economic and Technological Development Zone, Zhejiang

Applicant after: Hangzhou Zheng Xiang Medicine Co., Ltd.

Address before: American California

Applicant before: Hao Xiaolin

TA01 Transfer of patent application right
GR01 Patent grant
GR01 Patent grant
TR01 Transfer of patent right

Effective date of registration: 20220222

Address after: 210000 Zhongdan Ecological Life Science Industrial Park, no.3-1, xinjinhu Road, Jiangbei new district, Nanjing City, Jiangsu Province

Patentee after: Nanjing Zhengxiang Pharmaceutical Co.,Ltd.

Address before: 310000 2 A0101 room 452, 6 Avenue, Baiyang street, Hangzhou economic and Technological Development Zone, Zhejiang

Patentee before: Hangzhou Zheng Xiang Medicine Co.,Ltd.

TR01 Transfer of patent right